Studies towards the total synthesis of biological active...

257
Studies Towards the Total Synthesis of Biological Active γ-Butyrolactones Dissertation zur Erlangung des Doktorgrades der Naturwissenschaften Dr. rer. nat. an der Fakultät für Chemie und Pharmazie der Universität Regensburg vorgelegt von Andreas Schall aus Runding Regensburg 2007

Transcript of Studies towards the total synthesis of biological active...

  • Studies Towards the Total Synthesis of

    Biological Active γ-Butyrolactones

    Dissertation

    zur Erlangung des Doktorgrades der Naturwissenschaften

    Dr. rer. nat. an der Fakultät für Chemie und Pharmazie

    der Universität Regensburg

    vorgelegt von

    Andreas Schall

    aus

    Runding

    Regensburg 2007

  • 2

    Die Arbeit wurde angeleitet von: Prof. Dr. O. Reiser

    Promotionsgesuch eingereicht am: 25. Mai 2007

    Promotionskolloquium am: 26. Juni 2007

    Prüfungsausschuss: Vorsitz: Prof. Dr. H. Krienke

    1. Gutachter: Prof. Dr. O. Reiser

    2. Gutachter: Prof. Dr. B. König

    3. Prüfer: Prof. Dr. S. Elz

  • 3

    Der experimentelle Teil der vorliegenden Arbeit wurde unter der Leitung von Herrn Prof. Dr.

    Oliver Reiser in der Zeit von Oktober 2003 bis Mai 2007 am Institut für Organische Chemie

    der Universität Regensburg sowie in der Gruppe von Prof. Dr. Paul Hanson, University of

    Kansas, angefertigt,

    Herrn Prof. Dr. Oliver Reiser möchte ich herzlich für die Überlassung des äußerst

    interessanten Themas, die anregenden Diskussionen und seine stete Unterstützung während

    der Durchführung dieser Arbeit danken.

  • 4

  • 5

    for my family…

    “If you want to build a ship, don't drum up the men to gather wood,

    divide the work and give orders. Instead, teach them to yearn for the vast and endless sea.”

    Antoine de Saint-Exupéry (1900 - 1944)

  • Table of Content

    6

    Table of Content

    1. APPROACHES TO THE TOTAL SYNTHESIS OF BIOLOGICAL ACTIVE

    GUAIANOLIDES WITH A TRANS-ANNULATED LACTONE MOIETY 8

    1.1 INTRODUCTION 8 1.2 BIOSYNTHESIS OF GUAIANOLIDES 10 1.3 RACEMIC APPROACHES TOWARDS GUAIANOLIDES 15 1.4 STEREOSELECTIVE TOTAL SYNTHESIS OF GUAIANOLIDES 21 1.5 HEMI-SYNTHESIS STARTING FROM SANTONIN 28 1.6 CONCLUSIONS 35

    2. AIM OF THIS WORK 36

    2.1 CYNAROPICRIN - THE HERB PRINCIPLE OF ARTICHOKE 36 2.2 IXERIN Y - A GUAIANOLIDE SESQUITERPENE LACTONE GLUCOSIDE 37 2.3 RETROSYNTHETIC ANALYSIS OF THE TARGET COMPOUNDS 38

    3. SYNTHESIS OF CHIRAL ALLYLSILANES 40

    3.1 SYNTHESIS OF THE ENANTIOMERIC PURE CYCLOPENTENONE 41 3.2 SYNTHESIS OF THE CHIRAL ALLYLSILANES 44

    4. SYNTHESIS OF THE CYCLOPROPYLCARBALDEHYDE 47

    5. FORMATION OF THE ANTI-SUBSTITUTED LACTONE ALDEHYDE 50

    6. INVESTIGATIONS TOWARDS 5,6,5-RING SYSTEMS 53

    6.1 INTRAMOLECULAR CARBONYL-ENE REACTION 53 6.2 SMI2-PROMOTED RADICAL CYCLIZATION 55

    7. INVESTIGATIONS TOWARDS THE GUAIANOLIDE CORE SKELETON 56

    7.1 RADICAL CYCLIZATION APPROACH 56 7.2 RING CLOSING METATHESIS APPROACH 59 7.3 SYNTHESIS OF A 3X3 SCAFFOLD LIBRARY 65

  • Table of Content

    7

    8. TOWARDS CYNAROPICRIN AND IXERIN Y 73

    8.1 INVERSION OF THE C4-STEREOCENTER 73 8.2 INVESTIGATIONS ON EPOXIDATIONS 75 8.3 TAMAO-FLEMING OXIDATION 76 8.4 OXIDATION AT THE C8-POSITION 81 8.5 ELIMINATION REACTIONS 82

    9. STEREOSELECTIVE SYNTHESIS OF SMALL MOLECULE HAT INHIBITORS 88

    10. SUMMARY 94

    11. EXPERIMENTAL PART 97

    11.1 GENERAL 97 11.2 ABBREVIATIONS 99 11.3 SYNTHESIS OF CHIRAL ALLYLSILANES 100 11.4 SYNTHESIS OF THE CYCLOPROPYLCARBALDEHYDE 115 11.5 FORMATION OF THE ANTI-SUBSTITUTED LACTONE ALDEHYDE 119 11.6 RADICAL CYCLIZATION 121 11.7 PRECURSORS FOR RING CLOSING METATHESIS 124 11.8 RING CLOSING METATHESIS 131 11.9 SYNTHESIS OF A 3X3 SCAFFOLD LIBRARY 137 11.10 TOWARDS CYNAROPICRIN AND IXERIN Y 150 11.11 STEREOSELECTIVE SYNTHESIS OF GCN5 INHIBITORS 165

    12. APPENDIX 170

    12.1 NMR - SPECTRA 170 12.2 X-RAY DATA 231

    13. REFERENCES 244

  • Introduction

    8

    1. Approaches to the total synthesis of biological active guaianolides with a trans-annulated lactone moiety

    1.1 Introduction

    Guaianolides, consisting of a tricyclic 5,7,5-ring system represent a large subgroup of

    naturally occurring sesquiterpene lactones exhibiting significant biological activity.[1,2] Plants

    containing such compounds as the active principles have been used in traditional medicine

    throughout history for treating conditions ranging from rheumatic pains, increase of bile

    production to pulmonary disorders.

    Figure 1. Skeletal relationships.

    As the name itself indicates, the core structure of the guaianolides is derived from Guaiane, a

    natural product with a cis-fused 5,7-bicyclic hydroazulene ring-system (Figure 1). With only a

    few exceptions the hydroazulene core is also cis-fused in the 5,7,5-tricyclic carbon skeleton,

    while the γ-butyrolactone ring is trans-annulated in approximately 85% of all known

    guaianolides.[3]

    This interesting class of natural products shows a broad range of biological activity (Figure 2)

    stimulating the development of research towards their total synthesis. Although several

    strategies especially towards monocyclic γ-butyrolactones are reported to date,[4-10] only a few

    groups succeeded in the total synthesis of guaianolides.[10-14]

  • Introduction

    9

    H

    OR3

    OAc

    O

    R1OHOHO

    OH

    R2

    Thapsigargins (6)

    DehydrocostusLactone (2)

    OHH

    O

    H

    H OHH

    OO

    H

    Cladantholide (3)

    H

    Estafiatin (4)

    O

    O

    HHO

    H

    H

    Eremanthin (5)

    OHH

    O

    H

    H

    isolated fromcostus root (mokko)antimycobacterial activity(MIC = 2-16 µg/ml)

    isolated fromEremanthus elaeagnusschistosomicidal activity

    isolated fromCladanthus arabicusantifeedant activity

    isolated fromThapsia garganicapotent Ca-modulatingproperties

    O

    HO

    H

    H

    O

    Arglabin (1)isolated fromArtemisia glabellafarnesyl transferaseinhibitor(IC50 = 0.9-5.0 µg/ml)

    isolated fromArtemisia mexicanaantihelminthic activity

    Figure 2. Some guaianolides, representing the structural diversity of this class of compounds.

    The structure-activity relationship (SAR) of α-methylene sesquiterpene lactones was

    intensively studied.[15-20] It was shown that these compounds can react by conjugate addition

    of various biological nucleophiles such as cystein or thiol-containing enzymes (E-SH)

    (Scheme 1). Consequently, α-methylene sesquiterpene lactones are good alkylation agents

    manifesting their biological activity but also their cytotoxicity.

    Scheme 1. Michael addition on α-methylene sesquiterpene lactones.

    There is further evidence, that compounds of this type inhibit cellular enzyme activity and do

    not show DNA-alkylating properties.[18,21-26] Furthermore it is assumed that the residual

    substitution pattern of the guaianolides determines the specificity and the resulting biological

    activity.[11]

  • Introduction

    10

    1.2 Biosynthesis of guaianolides

    1.2.1 The mevalonate pathway

    Since ancient times various oils with intensive and mostly delightful fragrances were

    extracted from numerous plants. In the beginning direct distillation and later on steam

    distillation were common techniques to afford the essential oils, which mainly consisted of

    terpenes. Until now more than 30,000 terpenes from all sources have been identified, making

    them a large and structurally highly diverse family of natural products. It was early

    recognized that terpenes are formally derived from C5-isoprene units (7), but that isoprene (8)

    itself, a metabolite produced naturally, is not involved in their formation (Figure 3).

    Figure 3. Comparsion of C5-units.

    The biochemically active isoprene units are isopentenyl-pyrophosphate (IPP, 9) and

    γ,γ-dimethylallyl-pyrophosphate (DMAPP, 10). These important precursors are formed via

    certain biochemical pathways that have been extensively studied over the last 50 years leading

    to the generally accepted mevalonate (MVA) biosynthesis pathway of terpenes in

    organisms.[27-30] More recently a second biosynthetic route was discovered in plants also

    leading to IPP (9) and DMAPP (10) as the final products.[30-32] This so called mevalonate

    independent pathway or methylerythritol-phosphate pathway (MEP) is only found in a few

    plants and microorganisms. It was also recognized that the MVA-pathway is located in the

    cytosol and the MEP-pathway takes place in the plastids (chloroplasts, leukoplasts, etc.).

    Furthermore, in organisms equipped with both pathways, a limited exchange of intermediates

    between MVA and MEP also appears. This may explain why the MEP pathway was

    completely overlooked until labeling experiments revealed its existence.[33-36]

    The biosynthesis in the cytosol starts with the assembly of three molecules of activated acetic

    acid (acetyl-CoA) (11) by an initial Claisen-condensation and a subsequent aldol reaction to

    give β-hydroxy-β-methyl-glutaryl-CoA (13) (Scheme 2).

  • Introduction

    11

    CoAS CH3

    O CH3

    OCoAS CoAS

    O CH3

    O

    O

    SCoA

    -HSCOA+H2O

    O

    SCoA

    HO CH3HO2C OH

    HO CH3HO2C

    (NADPH + H+)

    -HSCoA

    +

    11 11 12

    11

    13Mevalonate (14)

    ATPOPP

    HO CH3HO2C

    -CO2

    -H2O

    OPP OPP

    IPP (9) DMAPP (10)

    15

    Scheme 2. MVA pathway for the biosynthesis of IPP (9) and DMAPP (10).

    Reduction with NADPH+H+ releases mevalonic acid (Mevalonate, MVA, 14), which is then

    activated by means of ATP to the pyrophosphomevalonate (15). Decarboxylation and

    elimination leads to isopentenyl-pyrophosphate (IPP, 9), further isomerization of the double

    bond to dimethylallylpyrophosphate (DMAPP, 10).

    To construct the basic backbones of terpenes, prenyltransferases connect IPP (9) and its

    isomer DMAPP (10) in a head to tail fashion (Scheme 3). In a first step DMAPP (10) is

    therefore ionized to an allylic cation 16, to which the double bond of IPP (9) can add resulting

    in a tertiary cation 17. Subsequent stereoselective loss of a proton introduces selectively a new

    trans substituted double bond and releases geranylpyrophosphate (GPP, 18), a fundamental

    precursor for the biosynthesis of monoterpenes (e.g. menthol).

  • Introduction

    12

    IPP (9)

    OPPOPP

    HSHR

    electrophilicaddition

    OPPHSHR

    stereospecificloss of proton

    OPP

    GPP (18)monoterpenes (C10)

    DMAPP (10) 16

    17

    OPPHSHR

    1. electrophilic addition

    OPP

    FPP (19)

    sesquiterpenes (C15),e.g. Guaianolides

    IPP (9)

    2. stereospecific lossof proton

    Scheme 3. Biosynthesis of GPP (18) and FPP (19).

    For the biosynthesis of sesquiterpenes the C10-skeleton of GPP (18) has to be extended by

    addition of a further C5-IPP (9) unit according to the isoprene rule[37-39] ((C5)n, n = 3 for

    sesquiterpenes) which was first discovered by Otto Wallach in 1887 but largely ignored until

    Leopold Ruzicka recognized its general significance. Repeating the electrophilic addition of

    IPP (9) and stereospecific elimination (Scheme 3) gives rise to farnesylpyrophosphate

    (FPP, 19), the precursor for linear and cyclic sesquiterpenes and sesquiterpene lactones.

    1.2.2 Biosynthesis of guaianolides

    The further assembly of guaianolides used in nature has been intensively investigated by

    de Kraker et al. based on the biosynthetic route of sesquiterpene lactones in chicory which is

    reasonable to assume to be also valid for other plant species (Scheme 4).[40-43] According to

    these studies, cyclization of FPP (19) yields (+)-Germacrene A (20). Because of the double

    bond configuration in FPP (19) two (E)-substituted double bonds are incorporated within the

    10-membered ring system of 20.

    Oxidation of the isopropenyl side chain by (+)-Germacrene A hydroxylase to the primary

    alcohol 21 and further oxidations by NAD(P)+-dependent dehydrogenases afford

  • Introduction

    13

    Germacrene acid (23). It was further demonstrated that hydroxylation on the C6-position and

    subsequent lactonization yields (+)-Costunolide (25).

    This intermediate is seen as a branching point in the biosynthesis of sesquiterpene lactones,

    because here the pathways for the formation of guaianolides, eudesmanolides and

    germacranolides are considered to divide.

    PPO

    FPP (19)

    PPO- NADPHO2

    NADP+H2O

    (+)-Germacrene A (20) NAD(P)+

    NAD(P)H

    NAD(P)+

    NAD(P)H

    HO2C

    Germacrene acid (23)

    21

    22

    NADPHO2

    NADP+H2O

    HO2CHOO

    O

    H2O

    24(+)-Costunolide (25)

    Guaianolides, e.g. Arglabin

    Eudesmanolides, e.g. Santonin

    Germacranolides, e.g. Parthenolide

    6 6

    H

    H

    H H

    OHC

    H

    HOH2C

    HHH

    3

    Scheme 4. Biosynthesis of (+)-Costunolide (25).

    Quite a number of stereospecific biomimetic transformations of germacranolides and their

    derivatives into eudesmanolides and guaianolides have been reported in literature.[44-52] Based

    upon these studies it is postulated that the second cyclization of germacranolides towards the

    guaianolide skeleton is directed by epoxidations or hydroxylations of the costunolide

    skeleton 25. Enzymatic epoxidation on C4-C5 position directly affords Parthenolide (26)

    (Scheme 5). This interesting germacranolide is a highly active antimigraine agent isolated

    from feverfew and magnolia and also shows anti-inflammatory and anti-tumor activities.[53-55]

    Trans-annular cyclization of the strained ring system in 26 and subsequent elimination

    completes the guaianolide skeleton 27.[56]

  • Introduction

    14

    Scheme 5. Guaianolides (27) via cyclizations starting from (+)-Costunolide (25).

    In addition to the above described route also an alternative pathway is proposed: Enzymatic

    introduction of a hydroxy group at C3-position in (+)-Costunolide (25) affords 28.

    Subsequent dehydration and cyclization also leads to the guaianolide skeleton 27.[43]

    Further oxidation steps on the 5,7,5-membered ringsystem of 27 introduce many different

    functionalities: Epoxides (e.g. found in Arglabin (1) or Estafiatin (4)) or the introduction of

    hydroxy groups (see Thapsigargins (6)) on various positions contributes to the diversity and

    complexity of this interesting and biologically important class of natural products.

    Esterification or glycosylation[57] of the later also broadens the structural variety of the

    guaianolides.

    In summary, nature has proven a tremendous creativity in the construction of the guaianolides

    with respect to their structures and biological functions. For an organic chemist now the

    question arises how to find synthetic entries towards these natural products. Even with

    modern state of the art techniques in organic synthesis at hand, the complexity of the core-

    structure and the high substitution pattern still makes the class of the guaianolides a

    challenging and exciting target.

  • Introduction

    15

    1.3 Racemic approaches towards guaianolides

    1.3.1 Total synthesis of (±)-Compressanolide and (±)-Estafiatin

    Although there are some reports in literature dealing with the synthesis of pseudo-

    guaianolides[1] or of guaianolide related compounds,[58-61] to the best of our knowledge the

    first total synthesis of a guaianolide with a trans-annulated lactone moiety was reported by

    Vandewalle et al.[62-64] in 1982. On the basis of a novel, flexible and convergent route towards

    substituted hydroazulenes the total synthesis of various sesquiterpene lactones has been

    achieved.[65]

    As the starting point the photochemical addition of 1,2-bis[trimethylsiloxy]-cyclopentene (29)

    to cyclopentenone (30) affords the 5,4,5-membered ring system (±)-31 as a single

    diastereomer, in which the five-membered rings are anti-oriented to each other (Scheme 6).

    OR

    OR

    h

    O

    OO

    OHH

    ORRO

    HH

    OHHO

    1. Ph3P=CH2

    Pb(OAc)4 p-TsOH,O O

    O O

    O

    2. HCl

    85%71%

    74%

    65%

    10

    [2+2]

    29 30 (±)-31

    (±)-32 (±)-33 (±)-34

    RO

    ROH

    H O

    R = TMS

    Scheme 6. Synthesis of key intermediate (±)-34.

    Subsequent Wittig-reaction and TMS-deprotection set the stage for ring expansion by

    oxidative cleavage of the diol (±)-32, giving rise to (±)-33 in which the exo-methylene double

    bond had concurrently migrated in conjugation to the carbonyl group. Acid catalyzed

    acetalization chemoselectively protected the more reactive carbonyl group on C-10 to afford

    the racemic key intermediate (±)-34, which allowed access to a number of different

    sesquiterpene lactones.

    With the substituted hydroazulene (±)-34 in hands the group of Vandewalle started out for the

    total synthesis of (±)-Compressanolide (44),[62] a guaianolide first isolated from Michelia

    compressa.[66] Furthermore a small variation of this route allowed the synthesis of

  • Introduction

    16

    (±)-Estafiatin (4),[62,64] a natural product first isolated by Romo and co-workers from

    Artemisia mexicana (Willd).[67]

    The opening step entails the epoxidation of the double bond in the 5-membered ring of (±)-34

    (Scheme 7, top). Controlled by steric hindrance (shielding of the β-face by the bulky ketal

    protecting group) the bulkiest reagent gave the best selectivity of 6:1 for the desired

    α-epoxide (±)-35.

    In contrast, epoxidation of the key intermediate (±)-34 using H2O2 afforded the epoxide in

    better yield and a 1:9 ratio, this time approaching from the β-face (Scheme 7, bottom) forming

    the now desired trans-fused 5,7-membered ring system (±)-36.

    Triton B,

    OOHPh1. LDA,

    2. separate3. DBU40%

    78%

    Br

    (±)-34

    OO

    O

    OO

    HO

    O

    OH

    7

    (±)-35 (±)-37

    7

    2:1

    1. H2O22. separate

    O O

    HO

    O

    7

    O O

    HO

    O

    71. LDA,

    Br

    70%

    = 6:1

    60%

    (±)-36 (±)-38

    4:1

    =1:9

    Scheme 7. Stereoselective epoxidation and alkylation.

    The next key step in the synthesis is the selective introduction of the prenyl-sidechain at the

    C7-position which is later on used for the formation of the lactone moiety. Attempts to

    introduce this arm by kinetic controlled deprotonation/alkylation already in the key

    intermediate (±)-34 failed, but alkylation of epoxide (±)-35 afforded the desired product

    (±)-37 in a 2:1 ratio (β:α). Base induced equilibration of the undesired epimer led again to an

    approx. 1:1 ratio, providing the possibility to recycle the unwanted epimer. Alkylation of

    (±)-36 on C-7 also introduced the prenyl-sidechain and afforded (±)-38 in a 4:1 ratio.

    The selective reductive opening of the epoxide in (±)-37 is the next crucial reaction, installing

    three stereocenters present in (±)-Compressanolide (44) within one step. This complex

    sequence starts with a reductive cleavage of the epoxide present in (±)-37 and a fast

    protonation of the resulting enolate (±)-39 (Scheme 8).

  • Introduction

    17

    H+

    R =

    OO

    HO

    OR

    O

    O

    HO

    OR

    HH

    O

    HO

    HO

    OR

    H

    (±)-37

    (±)-39 (±)-40

    1. Li, NH3, NH4Cl

    2. repeat57%

    HO

    HO

    HO

    O

    H

    (±)-41

    7

    Scheme 8. Stereoselective reduction sequence.

    Intramolecular tautomerization to ketone (±)-40 by intramolecular proton transfer from the

    near by hydroxy group leads to a less strained cis-annulated hydroazulene ring system.

    Subsequent in situ reduction gives rise to the more stable equatorial alcohol (±)-41, trans to

    the vicinal prenyl-sidechain at the C7-position.

    The trans-lactone moiety in (±)-42 is obtained by ozonolysis and Jones-oxidation of the

    prenyl-side chain completing the guaianolide skeleton (Scheme 9). Acid deprotects the

    masked ketone and a Wittig-reaction introduces the exo-methylenic double bond at the

    C10-position of (±)-43, a very common structural feature of sesquiterpene lactones.

    1. O3, DMS2. Jones 1. Ph3P=CH2

    1. LDA, MeI

    2. LDA, NH4Cl

    1:3(±)-Compressanolide (44)

    79%

    44%

    O

    OH

    HO

    HO

    H

    H

    O

    OH

    HHO

    H

    H

    (±)-42

    (±)-41 O

    OTMS

    HHO

    H

    H

    (±)-43

    3. HCl 2. TMSCl

    81 %

    10

    Scheme 9. Final steps towards (±)-Compressanolide (44).

    The resulting tertiary alcohol was protected, before α-methylation unfortunately afforded a

    mixture of 1:4 for the undesired isomer of (±)-44. Equilibration by kinetic protonation of the

    enolate improved the ratio only slightly to 1:3 towards (±)-Compressanolide (44).

  • Introduction

    18

    Applying the stereoselective reduction on (±)-38 as described above (Scheme 8) and sub-

    sequent oxidation gave rise to the all-trans substituted 5,7,5-ring system (±)-45 (Scheme 10).

    1. Li, NH3,NH4Cl

    2. O3, DMS

    3:1 endo:exo

    1. HCl2. separate

    3. Ph3P=CH2

    1. LDA, CH2O2. MsCl

    3. DBU4. mCPBA

    (±)-Estafiatin (4)(4.3% overall)

    56%

    3. Jones

    Burgess

    57%61%

    O

    O

    HHO

    H

    HOHH

    O

    H

    H

    OHH

    O

    H

    H

    OO

    OHH

    O

    H

    H

    OO

    OH

    75%

    (±)-45(±)-46

    (±)-47

    (±)-38

    Scheme 10. Finals steps in the synthesis of (±)-Estafiatin (4).

    The regioselective elimination of the tertiary alcohol present in (±)-45 proved to be difficult:

    As classical methods for the dehydration failed, only the application of Burgess-reagent

    resulted in a 3:1 endo:exo elimination towards (±)-46. Acidic deprotection of the ketal also is

    accompanied with an equilibration of the resulting ketone to 3:1 for the desired more stable

    cis-connected endocyclic alkene (±)-47. Subsequent Wittig-olefination and α-methylenation

    followed by epoxidation of the more reactive trisubstituted endocyclic double bond finally

    afforded (±)-Estafiatin (4) in 4.3% overall yield.

  • Introduction

    19

    1.3.2 Total synthesis of (±)-Dehydrocostus Lactone and (±)-Grosshemin

    Four years later Rigby et al.[68,69] reported the racemic synthesis of three further guaianolides

    ((±)-Dehydrocostus Lactone (2) (IC50 = 14 µM, CTL cells[70]), (±)-Estafiatin (4) and

    (±)-Grosshemin (62) starting from commercially available 2,4,6-cycloheptatrienone

    (Tropone) (48). Similar to the Vandewalle-approach described above, the first target was also

    the construction of the hydroazulene core. Utilizing the 7-membered ringsystem already

    present in Tropone (48), 1,8-addition of appropriate nucleophiles afforded the alkylated

    products 49 and 50, which were further converted into the aldehyde 51 and the

    diazocompound 52, respectively (Scheme 11).

    O

    BrMg O

    OOMe

    CHO

    Tropone (48)OLi

    OtBu

    O OPivO

    N2

    O

    OtBu

    O

    O O

    3 steps

    5 steps

    49

    50

    51

    52

    96%

    90%

    73%

    70%

    Scheme 11. Functionalization of Tropone (48) by Rigby et al.

    The required cis-fused hydroazulene ring system is then formed via a Lewis-acid mediated

    cyclization on 51 and subsequent reductive opening of the resulting epoxide (±)-53 releasing

    the next key-intermediate (±)-54 in the synthesis of (±)-Estafiatin (4) (Scheme 12, top).

    BF3 OEt2

    Cu/CuSO4

    H

    H

    O

    OMe

    O

    OPivHH

    1. Ac2O,BF3 OEt2

    OAc

    OPivH

    HAcO

    MEMO

    H

    H

    OMe

    1. Li, MeNH2

    2. MEMCl51

    52

    (±)-53 (±)-54

    (±)-55 (±)-56

    75% 93%

    92%

    80%

    Scheme 12. Formation of cis-fused hydroazulene systems.

  • Introduction

    20

    Alternatively an intramolecular cyclopropanation in 52 gives rise to the tricyclic system

    (±)-55 which is then opened by a Lewis-acid mediated homoconjugate addition reaction

    releasing the intermediate (±)-56 for the synthesis of (±)-Grosshemin (62) (Scheme 12,

    bottom).

    To introduce the trans-fused lactone moiety, the double bond in (±)-54 was first selectively

    epoxidized (approach of peracid from the less hindered upper face) and then the epoxide was

    opened with the appropriate lithium-organyle closing the ring to lactone (±)-57 (Scheme 13).

    Functional group transformation leads in 4 steps to the diene (±)-58.

    1. mCPBA

    2.

    OLi

    OLi OHH

    MEMO

    O

    OMe

    1. LDA,Me2NCH2I

    2. MeI

    (±)-DehydrocostusLactone (2)

    (1.0% overall)

    1. BF3 OEt2

    2. mCPBA

    (±)-Estafiatin (4)(0.3% overall)

    4 steps(±)-54

    H

    H

    OHH

    OH

    H

    OHH

    OH

    H

    OHH

    OH

    H

    O

    (±)-57 (±)-5842%

    71% 35%

    6%

    Scheme 13. Final steps towards (±)-Dehydrocostus Lactone (2) and (±)-Estafiatin (4).

    The still missing α-exo-methylenic group is introduced via a Mannich-type reaction to yield

    (±)-Dehydrocostus Lactone (2) in 1.0% overall yield. The structural closely related

    (±)-Estafiatin (4) was then obtained in 0.3% overall yield by selective isomerization of the

    double bond present in (±)-2 towards the more stable tetrasubstitution and subsequent regio-

    and stereoselective epoxidation.

    Using the hydroazulene key intermediate (±)-56 Rigby and coworkers started out for the

    synthesis of (±)-Grosshemin (62), first isolated by Rybalko et al. from Grossheimia

    macrocephala.[71] The methyl group present in (±)-Grosshemin (62) was selectively

    introduced via alkylation by methyliodid approaching over the less hindered upper face of

    (±)-56 (Scheme 14).

  • Introduction

    21

    1. MeLi, MeI

    2. TMSOTf,(TMSOCH2)2 O

    OPivH

    HAcO OO

    H

    HHO O

    1. VO(acac)2,tBuOOH

    2. LiCH2CO2Li

    O

    HH

    OO

    HO

    O

    1. LDA,Me2NCH2I

    2. MeI

    3. H+HH

    O

    HO

    O

    O(±)-Grosshemin (62)

    (8.6 % overall)

    6 steps(±)-56

    H

    H

    H

    H

    (±)-59 (±)-60

    (±)-61

    56%

    76%

    65% 71%

    Scheme 14. Final steps towards (±)-Grosshemin (62).

    Further six steps including functional group transformation and Wittig olefination lead to

    allylalcohol (±)-60. The trans-fused lactone ring is introduced by directed epoxidation via the

    allylic alcohol and subsequent epoxide opening as shown in (±)-61. Again a Mannich-type

    reaction introduced the α-exo-methylene group at the lactone ring and finalized the synthesis

    of (±)-Grosshemin (62) in 8.6% overall yield.

    1.4 Stereoselective total synthesis of guaianolides

    1.4.1 Total synthesis of (+)-Cladantholide and (-)-Estafiatin

    A very elegant stereoselective approach towards two members of the guaianolide family via a

    radical cyclization cascade was reported by Lee and co-workers,[72] who succeeded in the total

    synthesis of (+)-Cladantholide (3) (isolated from Cladanthus arabicus (L.) Cass.)[73] and

    (-)-Estafiatin (4) starting from (R)-Carvone (63).

    In three steps the chlorohydrin derivative 64 was synthesized, which was subjected to a

    stereoselective Favorskii-rearrangement to afford the highly substituted cyclopentane-

    carboxylate 65 (Scheme 15). Three more steps were necessary to obtain the bromoacetal 66,

    which was set up for a radical cyclization being initiated by AIBN/Bu3SnH under standard

    high-dilution conditions. 67 was obtained in quantitative yield and perfect diastereoselectivity

    with respect to the newly created stereocenters at the C7- and the C10-position.

  • Introduction

    22

    O

    H3 steps

    O

    H

    ClOTHP

    H MeO2CH

    OTHP

    (R)-Carvone (63) 64 65

    Favorskii-rearrangement

    58% from 63

    H

    OTHPHO

    Br

    EtO OHH

    OTHP

    EtO7

    10AIBN,Bu3SnH

    H

    H

    66 67

    75% 99%

    3 steps

    H

    Scheme 15. Favorskii-rearrangement and radical cyclization.

    The stereochemical outcome of this highly selective and efficient cascade can be explained by

    conformational analysis of the substrate (Scheme 16): The most stable conformation of the

    cyclopentane ring in 68 is represented with three substituents in equatorial positions, and the

    attached sidechains are oriented chairlike.

    Scheme 16. Conformational analysis of 68 and radical cyclization.

    Consequently 5-exo-cyclization of the primary radical onto the opposite double bond forms

    the trans cyclic acetal 69 setting the correct stereochemistry on C7. Subsequent 7-endo

    cyclization affords the tertiary radical 70, while the alternative kinetically favored 6-exo

    pathway was not observed. Final hydrogen addition setting the correct stereocenter at C10

    must have occurred from the α-face, which is presumable sterically less hindered.

    The high preference for the 7-membered ring formation by radical cyclization was also

    observed by Reiser and co-workers during their investigations on model systems towards the

    synthesis of bi- and tricyclic sesquiterpene lactones of the xanthanolide and guaianolide

    family (Scheme 17).[74]

  • Introduction

    23

    OO

    Br

    CO2EtH

    H

    Bu3SnH/AIBN

    7-endoOO

    CO2EtH

    H

    71 72

    83-95%

    Scheme 17. 7-endo cyclization by Reiser et al.

    To finalize the synthesis of (+)-Clandantholide (3) Lee et al. had to transform 67 to the

    ketoacetal 73, in which the introduction of a hydroxy group adjacent to the ketogroup yields

    α-hydroxyketone 74 (Scheme 18).

    1. LDA,TMSCl

    2. Dimethyl-dioxirane

    1. TsNHNH2

    OHH

    OO

    H

    (+)-Cladantholide (3)H

    OHH

    OHMeO

    H

    H

    O

    OHH

    MeO

    H

    H

    O

    67

    73 74

    80%

    79%

    OHH

    OHMeO

    H

    H

    2 steps

    2. MeLi

    40%

    46%

    2 steps

    75

    OHH

    N

    MeO O

    HN

    Li

    Ts

    H

    H

    76

    Scheme 18. Synthesis of (+)-Cladantholide (3).

    Applying the Shapiro-protocol to 74 resulted in the regioselective introduction of the C=C-

    double bond to give the allylalcohol 76. It was argued, that the reaction proceeds through an

    intermediate 75, in which the lithium coordinates with the first nitrogen of the hydrazone and

    the adjacent hydroxy group. Consequently, excess base can only deprotonate next to the

    methyl group, affording the trisubstituted double bond in 76. Finally, oxidation and

    stereoselective α-methylation completed the synthesis of (+)-Cladantholide (3).

  • Introduction

    24

    H

    OTHPH

    O

    Cl

    O OHH

    OTHP

    O7

    10

    H

    H

    77 78

    65%

    65

    MeO2C MeO2C ClMn(OAc)3Cu(OAc)2

    86%

    2 steps

    OHH

    OH

    HMeO2C2 steps

    71%

    (-)-Estafiatin (4)

    O

    O

    HHO

    H

    H

    79

    1. LiOH, CH2NMe2I

    2. mCPBA54%

    Scheme 19. Synthesis of (-)-Estafiatin (4).

    65 has also been the starting point for the synthesis of (-)-Estafiatin (4) (Scheme 19), for

    which again a radical cascade cyclization has been the key step. In difference to the reductive

    conditions employed for the transformation of 66 to 67, the cyclization of 77 to 78 was carried

    out under oxidative conditions, being initiated by hydrogen rather than halogen abstraction of

    the α-halo-carbonyl functionality. Reductive dechlorination and dehydration proceeded

    uneventfully to 79 and subsequent α-methylenation using Eschenmoser´s salt and selective

    epoxidation of the endo double bond afforded (-)-Estafiatin (4).

    1.4.2 Synthesis of the Thapsigargins by Ley et al.

    A powerful demonstration of modern organic synthesis was shown by Ley and co-workers

    with their total synthesis of the Thapsigargins (6).[75-77] Although extracts from the root of

    Thapsia garganica L. were used for a long time as treatment for rheumatic pains and

    pulmonary disorders, the identification and characterization of the active principles was not

    reported until 1980.[78,79] The potent biological activities reach from histamine liberation[80] to

    selective Ca2+-modulating properties[81-83] on subnanomolar concentrations.

    The outstanding activity and the complex molecular structure consisting of a polyoxygenated

    5,7,5-core structure which is further functionalized with eight stereogenic centers and up to

    four different ester groups, makes this class of guaianolides to an especially challenging target

    for total synthesis.

    The overall strategy towards the Thapsigargins (6) was again to construct the hydroazulene

    core first and subsequently functionalize it towards the target.

  • Introduction

    25

    Therefore Ley and co-workers started from (S)-Carvone (63) following a similar route as

    described above for Lee et al.[72] Within five steps 80 was reached and further eight high

    yielding steps lead to aldehyde 81 with already one allyl sidearm installed (Scheme 20).

    Scheme 20. Ring closing metathesis as an essential key step.

    The second arm for the ring closing metathesis key step was introduced using the lithium

    anion of ethylvinylether. Following strictly the Felkin-Anh model diene 82 was generated as a

    single diastereomer. The bicyclic hydroazulene ringsystem was then constructed by ring

    closing metathesis affording 83 in high yield.

    The convex half space of the double bond present in 83 is shielded by the bulky TES

    protecting group and so osmylation results in good selectivity (16:1) for the concave attack

    releasing 84 (Scheme 21).

    Scheme 21. Facial selective osmylation.

  • Introduction

    26

    Esterification of the resulting alcohol and subsequent intramolecular Horner-Wadsworth-

    Emmons reaction provided the butenolide 85 which was within 3 steps transformed into 86.

    Further functionalization of 86 towards the highly oxygenated core system of the targets was

    performed by selective dihydroxylation of the side chain to yield 87 (Scheme 22). The desired

    trans-annulated lactone 88 was formed after selective oxidation of the primary alcohol.

    Scheme 22. Dihydroxylation and completion of the tricyclic framework.

    Until this point already 23 steps were necessary, but this process required five

    chromatographic steps only, providing a nice possibility to assemble material in 11% overall

    yield to this point in multiple gram quantities. After MOM deprotection the acetonide 89 was

    formed stabilizing the already complex system.

    The next target was the modification of the cyclopentane ring. Kinetic enolisation of 89

    followed by oxidation from the less hindered concave face provided α-siloxy-ketone 91

    (Scheme 23).

  • Introduction

    27

    HH

    OR

    OR

    O

    OROO

    OHH

    O

    OR

    O

    OROO

    O OR

    Dimethyldioxirane,acetone

    R = TMS

    HH

    OR

    OR

    O

    OROO

    O OR

    TMSCl,NEt3

    H

    O

    OR

    O

    OROO

    O

    PhSeBr

    TMSCl,NEt3

    HH

    HH

    89

    90 91

    92 96

    88% 99%

    90% 94%

    H

    H

    OTMS

    OSePh

    H

    H

    OTMSSe+

    Ph

    -O

    H

    OTMS

    93 94 95

    Scheme 23. Synthesis of conjugated ketone by selenium elimination.

    Again, enolisation, this time to the opposite side, afforded highly functionalized 92, the

    starting point for a complex unanticipated catalytic selenium reaction. The opening step in

    this sequence is supposed to be a selenation of the TMS protected secondary alcohol 92,

    because this seems to be the least hindered position. Subsequent 2,3-sigmatropic

    rearrangement affords a selenoxide 94 which is able to undergo syn-elimination in direction

    towards the 7-memberd ringsystem. Hydrolysis of the resulting enolether 95 releases the

    conjugated ketone 96.

    To complete the synthesis Ley and co-workers still had to set the last stereocenter in the

    cyclopentane ring of 96 by stereoselective reduction (4:1 selectivity, again by steric controlled

    attack from the concave face). Esterification with angelic acid of the resulting alcohol and

    removal of the TMS protecting groups afforded the diol 97 (Scheme 24). Selective acetyl

    protection of the more reactive hydroxy group via a polymer supported reagent installed the

    second ester group in 98.

  • Introduction

    28

    H

    O

    OH

    O

    OHOO

    O

    1. NaBH42. angelic acid, NEt3,

    trichlorobenzoylchloride

    O3. TBAF

    1. isoprenylacetate,polymer-supported-TsOH

    R =

    O

    Nortrilobolide 72%O

    Trilobolide 78%O

    Thapsivillosin F 73%

    R2O,DMAP H

    O

    OAc

    O

    ROHOHO

    OOH

    H

    O

    OAc

    O

    HOHOHO

    OOH

    H96

    97

    98 99

    65% 68%2. HCl (aq.), MeOH

    4 4

    Scheme 24. Final synthetic steps towards the Thapsigargins.

    Removal of the acetal protection provided the triol 98. Esterification of the more reactive

    secondary alcohol on the C-4 position finally afforded three members of the Thapsigargin

    family.

    1.5 Hemi-Synthesis starting from Santonin

    First isolated by Kahler et al. in 1830 [84,85] it was a long and exciting way to elucidate the full

    structure of (-)-α-Santonin (100).[85-90] Commercially available by extraction,[91] this

    eudesmanolide provides a perfect starting point for the synthesis of various sesquiterpene

    lactones (Scheme 25). Abe et al.[92-94] and Marshall et al.[95] also succeeded in the total

    synthesis starting from a hexahydronaphtalene skeleton or 3-methyl-benzoic acid,

    respectively.

  • Introduction

    29

    Scheme 25. Structure and rearrangement of (-)-α-Santonin (100).

    Despite its own biological activity, the most important feature of (-)-α-Santonin (100) is the

    possibility to rearrange the 6,6,5-eudesmanolide skeleton to a hydroazulene carbon backbone

    (Scheme 25). The light induced rearrangement of 100 is one of the longest known

    photochemical organic reactions.[96] The cross conjugated dienone rearranges upon irradiation

    in the presence of acetic acid towards acetyl-isophotosantonic lactone 101 and serves as a

    classic example for photochemical rearrangements, although it was a long way to completely

    understand this reaction.[85,96-99] Furthermore was found that solvolysis of methanesulfonates

    102 also provides an entry to the 5,7,5-ringsystem of the guaianolides.

    1.5.1 Syntheses by Ando et al.

    Ando et al. were able to synthesize more than ten guaianolides starting from (-)-α-Santonin

    (100). Preparing suitable derivatives of this available natural product and subsequent

    solvolytic rearrangement offers a very interesting and efficient entry towards the guaianolides.

    1.5.2 Synthesis of (+)-Arborescin

    (+)-Arborescin (107) was first isolated by Meisels and Weizmann from Artemisia arboresces

    (Compositae), a plant used for contraceptive purpose by the ancient Greeks and Arabs.[100]

    The proposed structure by Herout et al.[101] was later on confirmed by X-ray analysis.[102]

  • Introduction

    30

    In an opening step Ando et al.[103] transformed (-)-α-Santonin (100) into the eudesmanolide

    103 (Scheme 26). Protecting group transformation and reduction afforded the alcohol 104.

    mCPBA

    OO O

    OH

    OH

    1. BzCl2. H3O+

    3. Zn(BH4)2

    50%

    O

    OBz

    HO

    1. MsCl

    2. AcOK63%

    22%O

    OBz

    HO

    O

    O

    HO

    O1. K2CO3 / H2O2. MsCl

    3. Li2CO3 / LiBr(+)-Arborescin (107)

    31%

    H

    H OO

    OHH

    OBz

    H

    H

    H

    H

    H

    H

    H

    H

    103 104

    105 106

    100

    Scheme 26. Synthesis of (+)-Arborescin (107).

    Mesylation and solvolytic rearrangement resulted in a mixture of olefins 105, which was

    rectified by selective epoxidation of the tetrasubstituted double bond of the endo-isomer. The

    approach of the epoxidizing agent from the sterically less hindered down face sets the right

    stereochemistry for the epoxide. Subsequent deprotection and elimination afforded

    (+)-Arborescin (107) with a trans annulation of the cyclopentane ring.

    1.5.3 Synthesis of Zaluzanines

    A similar approach was used for the synthesis of different Zaluzanines (111).[104,105] These

    guaianolides were originally isolated from Zaluzania augusta and Zaluzania triloba[106,107]

    and show high biological activities for example in tumor inhibition.[108]

    The rearrangement of the (-)-α-Santonin (100) derivative 108 to the guaianolide skeleton 109

    resulted again in a mixture of double bond isomers which was rectified by selective

    epoxidation of the endo-isomer (Scheme 27).

  • Introduction

    31

    OO

    O

    OMsH

    PPh3, DEAD,AcOH

    O

    OR

    HO

    H

    111R = H Zaluzanin CR = Ac Zaluzanin D

    100 O

    OH

    HO

    H1. KOAc2. K2CO3

    O

    OAc

    HO

    H

    46%

    44%

    3 steps

    H

    H

    H

    H

    H

    H

    H

    H

    108 109

    110

    Scheme 27. Synthesis of (+)-Zaluzanin C/D (111).

    Subsequent Mitsunobu-inversion of the secondary alcohol in 109 sets the right

    stereochemistry of the hydroxygroup in the cyclopentane ring of 110 and afforded after

    introduction of the exo-methylenic bond (+)-Zaluzanin C and D (111).

    1.5.4 Mokko lactone, Dehydocostus Lactone and Eremanthin

    Mokko Lactone (114) and Dehydrocostus Lactone (2) (both isolated from costus root

    (mokko)[109,110]) and Eremanthin (5) (isolated from the hartwood oils of Eremanthus

    elaeagnus and Vanillosmopsis erytrhoppa)[111-113] are also accessible via this route. A

    common feature of these three natural products is the lacking of the hydroxy functionality in

    the cyclopentane ring.

    Starting again from (-)-α-Santonin (100) Ando and co-workers synthesized 112 (Scheme 28).

    After deprotection of the ketal and double bond isomerization the resulting ketone was

    reduced to the secondary alcohol 113, which is needed for subsequent mesylation. Solvolysis

    directly affords Mokko Lacone (114) accompanied with its double bond isomers 115 and 116.

    Desaturation of 114 and 115 releases Dehydrocostus Lactone (2) and Eremanthin (5),

    respectively

  • Introduction

    32

    O

    HO

    O

    O

    1. AcOH / H2O2. Br23. Zn-Hg4. LiAl(OtBu)3H

    40%

    O

    H OHO 6:1

    1. MsCl

    2. KOAc OHH

    O

    Mokko Lactone(114)

    + +

    81%

    Dehydrocostus Lactone(2)

    Eremanthin(5)

    1. LDA, (PhSe)22. H2O2

    H

    H H

    H100

    H

    H OHH

    O

    H

    H O

    HO

    H

    H

    OHH

    O

    H

    H OHH

    O

    H

    H

    112 113

    115 116

    Scheme 28. Further Guaianolides starting from (-)-Santonin (98).

    This strategy allowed Ando et al. to succeed in the total synthesis of over ten different

    guaianolides (Figure 4).[114]

    OH

    OH

    H

    OHH

    OH

    H

    Dehydrocostus LactoneIsodehydrocostus Lactone

    OHH

    OH

    H

    O

    2-Oxodesoxyligustrin

    OH

    OH

    H

    O

    LeucodinDehydroleucodin

    OH

    OH

    H

    O

    Ludartin11 ,13-Dihydroludartin

    OOHH

    OH

    H

    8-Deoxy-11 ,13-dihydrorupicolin B

    Kauniolide11 ,13-Dihydrokauniolide

    Figure 4. Some guaianolides prepared by Ando and co-worker.

  • Introduction

    33

    Inspired by this route Pedro et al. were also able to present stereoselective hemi-syntheses for

    (+)-11βH,13-Dihydroestafiatin, (+)-11βH,13-Dihydroludartin, (-)-Compressanolide (44), and

    (-)-11βH,13-Dihydro-micheliolide starting from (-)-α-Santonin (100).[115]

    1.5.5 Biomimetic synthesis of Absinthin

    Several short biomimetic syntheses of several guaianolides starting from suitable modified

    natural germacranolides have also been reported in literature.[47,49,51,52] The main problems

    within these approaches are the insufficient availability of the staring materials or the

    frequently observed complex mixtures during the cyclization reactions in combination with

    poor yields. For example Gallicin (117) a germacranolide isolated from Artemisia maritima

    gallica ssp Willd can be mesylated and subsequent cyclization affords the guaianolide

    skeleton which can be further converted into Compressanolide (44) (Scheme 29).

    Scheme 29. Biomimetic cyclization of germacranolides.

    Isolated in 1953 by Herout et al.[116-118] as a main dimeric guaianolide from Artemisia

    absinthium L. the complex structure of (+)-Absinthin (122) was not determined before the

    1980s.[119-122] The challenging structure and the biological activity of this compound inspired

    Zhang and co-workers to search for a synthetic approach towards this compound.[123]

    Photochemical rearrangement of (-)-α-Santonin (100) provided access to the guaianolide

    skeleton and reduction yielded the alcohol 118 (Scheme 30). Subsequent Mitsunobu-

    arylselenation followed by oxidative elimination afforded the precursor diene 119.

  • Introduction

    34

    HOAc, h

    NaBH4 OO

    H

    OAcH

    OH

    ArSeCN, PBu3

    OOH

    OAcH

    NaIO4

    1. neat, 10 d

    O

    O

    HHO

    H O

    O

    H

    OH

    HH

    HH

    H2. KOH

    H

    H

    [4+2]Cycloaddition

    HH100

    39% 48%

    72%

    118 119

    120

    1. SOCl2, NEt32. OsO4, NMO MeLi

    (+)-Absinthin (122)(18.6% overall)

    O

    O

    H

    O

    O

    H

    HH

    H

    OO

    H H O

    O

    H

    H O

    O

    H

    H

    HH

    H

    HOOH

    77%89%

    121

    3. NaIO4

    Scheme 30. Biomimetic dimerisation via [4+2] Cycloaddition.

    The biomimetic dimerisation of the two identical Diels-Alder partners proceeded highly

    regio- and stereospecific towards 120. This can be explained by minimizing the steric

    interactions during the approach of the reaction partners via the less hindered face (for the

    cyclopentadiene moieties). A head to head orientation (for the lactone moieties) also

    minimizes steric interactions between the 7-membered ring systems (inset box in Scheme 30).

    After basic cleavage of the acetyl-protecting groups the stereocenters of the resulting tertiary

    alcohol 120 had to be inverted. This was achieved by initial elimination and subsequent

    oxidative cleavage of the resulting exo-cyclic double bond to release diketone 121. To

    complete this biomimetic total synthesis of (+)-Absinthin (122), stereoselective methylation

    of the carbonyl groups afforded the target compound in 18.6% overall yield.

  • Introduction

    35

    1.6 Conclusions

    The search for new synthetic ways towards the guaianolides did not only result in new total

    synthesis of complex and biological active natural products, but also contributed to the

    development of a wide range of new and modern chemistry. The fundamental and

    methodological aspects of natural product synthesis have always proven to be of great

    importance beside the straight forward synthetic pathway towards the target structures. As

    there are more and more members of the guaianolide family discovered and extracted from

    different plants, the full evaluation of their biological activity is still of current interest.

    Therefore total synthesis has to find new, efficient and flexible ways to make these

    compounds and their derivatives available.

  • Aim of this work

    36

    2. Aim of this work This work seeks to explore a new synthetic strategy towards the total synthesis of two

    interesting members of the guaianolide family, namely Cynaropicrin (123) and Ixerin Y

    (126). A highly promising biological activity in combination with their complex structure

    makes these compounds challenging targets for total synthesis.

    2.1 Cynaropicrin - the herb principle of artichoke

    The artichoke (Cynara scolymus L.) was elected as the medical plant of the year 2003.[124]

    This 1.5-2 m tall perennial thistle is originated in southern Europe especially around

    Mediterranean (Figure 5).[125-129]

    Figure 5. Pictures of Cynara scolymus L. and structure of Cynaropicrin (123).

    Dried or fresh leaves and stems of Cynara are used as a choleretic (to increase bile

    production) and to treat gallstones. Furthermore, drugs are prepared from the extracts for the

    treatment of dyslipidemias, arteriosclerosis and inflammatory bowel disorders.[130]

    Responsible for the bitter taste of this vegetable is the guaianolide Cynaropicrin (123),[131,132]

    also identified as the active principle, which shows a broad variety of biological activities:

    Significant cytotoxicity against human tumor cell lines (ED50 = 0.23-1.72 µg/ml) was found

    by Choi and co-worker.[133] Further reports are provided in literature on the pro-apoptotic

    activity of Cynaropicrin (123) on leukocyte cancer cell lines[134] in combination with activity

    in acute and chronic inflammatory processes.[135-137] In addition to this, antibacterial effects

    are seen by irreversible inhibition of MurA, an enzyme responsible for the first step in

    cytoplasmatic biosynthesis of peptidoglycan precursors.[138] This enzyme is of certain interest

    for drug development projects, since the MurA-dependent metabolites are of vital importance

    for bacteria.[139-141]

  • Aim of this work

    37

    2.2 Ixerin Y - a guaianolide sesquiterpene lactone glucoside

    Various interesting natural products have been isolated from Ixeris plants (Figure 6), and

    sesquiterpene lactones as the Ixerins A-Z have been characterized (Figure 7).[142-146]

    Figure 6. Pictures of Ixeris denticulata f. pinnatiparita.

    A wide spectrum of biological activities, such as cytotoxicity[147] as well as having ant

    repellent[148] and antifeedant[149] properties is reported for some members of the Ixerin family.

    In addition to this, Ixerin Y (126) shows a promising inhibitory effect against the growth of

    human breast cancer cell lines (MCF7: IC50 = 6.36 µg/ml, MDA468: IC50 = 11.87 µg/ml).[57]

    Prominent members of the Ixerin guaianolide family are given in Figure 7.

    OOH

    Ixerin Z (127)

    H

    H

    O- -D-Glc

    OOO

    H

    H

    HO

    H

    Glc- -D-O

    Ixerinoside (128)

    OO

    R

    H

    H

    H

    O- -D-Glc

    125 R = -OH Ixerin X126 R = -OH Ixerin Y

    OOH

    H

    H

    O- -D-Glc

    H

    OH

    Ixerin D (124)

    4

    Figure 7. Prominent members of the Ixerin guaianolide family.

    A common feature of the mentioned members of the Ixerins is the glycosidic linkage of a

    glucose moiety found at different positions on the hydroazulene core skeleton. Ixerin X (125)

    and Y (126) differ only in the configuration of the hydroxy functionality at the C4-position

    which is absent in Ixerin D (124) and Z (127).

  • Aim of this work

    38

    Table 1. Sources of members of the Ixerin family.

    Entry Guaianolide isolated from Ref.

    1 Ixerin D (124) Ixeris tamagawaensis [143]

    2 Ixerin X (125) Ixeris Denticulata f. pinnatipartita and

    Ixeris sonchifolia resp.

    [57,150,151]

    3 Ixerin Y (126) Ixeris Denticulata f. pinnatipartita and

    Ixeris sonchifolia resp.

    [57,150]

    4 Ixerin Z (127) Ixeris Denticulata f. pinnatipartita and

    Ixeris sonchifolia resp.

    [150,152]

    5 Ixerinoside (128) Ixeris sonchifolia [153]

    The complex structure in combination with their broad spectrum of biological activity makes

    all these compounds to challenging targets for total synthesis.

    2.3 Retrosynthetic analysis of the target compounds

    All total synthetic approaches described so far (see introduction) find their first target in the

    construction of the guaiane core system. Once this skeleton is assembled, the trans-annulated

    lactone moiety is introduced in the synthesis. The stereochemistry is mostly substrate

    controlled via the hydroazulene core system and the target structures are finally reached by

    functional group transformations.

    In contrast to this, the synthetic approach towards the two guaianolides described above seeks

    for a different route: The lactone moiety and its trans-substitution pattern is constructed first

    as seen in lactone aldehyde 129, and the guaianolide skeleton is finalized by closing the

    hydroazulene substructure (Figure 8).

    Although Cynaropicrin (123) and Ixerin Y (126) have been isolated from very different

    sources, there seems to exist quite an interesting structural relationship between these

    compounds: The basic framework of 123 and 126 differs only in the position of the double

    bonds within the guaiane skeleton as well as in the position of the hydroxy group on the

    allylic moiety in the cyclopentane ring.

  • Aim of this work

    39

    Cynaropicrin (123)

    OO

    H H

    RO

    OH

    OO

    HO

    O- -D-Glc

    H

    R= OH

    O

    Mannichallylation

    ring closingmetathesis

    OO

    CHOHX

    OPgSiMe2R

    H

    H

    +

    E = CO2Me

    129 X = O, CH2R = Ph, OiPr

    (-)-130

    OPgSiMe2R

    X

    132 X = O, CH2R = Ph, OiPr

    Ixerin Y (126)

    H

    H

    H

    H

    Mannichallylationradicalcyclization

    TMS

    O E

    O

    OPg

    CO2Et

    CHOE(O)CO

    131

    133

    Figure 8. Retrosynthesis of Cynaropicrin (123) and Ixerin Y (126).

    Because of this, a stereoselective synthesis of both natural products is envisioned to the

    common precursor 129 using either a radical cyclization or ring closing metathesis approach

    to form the central seven membered ring.

    The lactone aldehyde intermediate 129 having already incorporated five stereocenters with

    respect to the target structures, was envisaged to be assembled from the cyclopropyl-

    carbaldehyde (-)-130, accesible from furan 131, and the disubstituted allylsilane 132. The

    construction of the latter is acchieved starting from the enantiomerically pure protected

    cyclopentenon 133.

  • Main Part

    40

    3. Synthesis of chiral allylsilanes Allylsilanes have proven to be versatile tools in organic chemistry, especially for the mild and

    highly selective Hosomi-Sakurai allylation.[154-158] To construct the southern cyclopentane

    ring of the guaianolides, two chiral allylsilanes with all the desired substituents in place were

    prepared. Compounds 134 and 135 are envisioned as important key intermediates with three

    major features (Figure 9):

    i) the allylsilane moiety allows the smooth addition onto the cyclopropylcarbaldehyde.

    ii) the protected alcohol already possesses the right stereochemistry (with respect to

    Cynaropicrin (123)) and can be further functionalized.

    iii) the bulky silyl group in the sidechain acts as a directing group during the addition

    reaction and can be transformed into a free hydroxy functionality later on.

    OPgSiMe2R

    TMS

    134 R = Ph135 R = OiPr

    O

    OPg

    OOH

    Furfuryl alcohol(136)

    133

    allylic system(i)

    stereocontroland

    functionalization(iii)

    functionalization(ii)

    Figure 9. Retrosynthesis of chiral allylsilanes 134 and 135, respectively.

    To be able to introduce all of these features, the synthesis of the allylsilanes 134 and 135 is

    referred to the chiral protected cyclopentenone 133, which is readily accessible in

    enantiomeric pure form, starting from inexpensive commercially available furfuryl

    alcohol (136).

  • Main Part

    41

    3.1 Synthesis of the enantiomeric pure cyclopentenone

    There are quite a number of methods known for the synthesis of optically active cis-2-

    cyclopenten-1,4-diol derivatives,[159-168] but many of these show certain inconveniences

    (e.g. cracking of dimer when starting from cyclopentadiene, instability of intermediates, loss

    of stereoselectivity during the synthesis).[169] To avoid these problems it was decided to

    follow a well established route reported by Curran et al. for large quantity preparation of

    optically active cis-2-cyclopenten-1,4-diols.[169,170]

    First, furfuryl alcohol (136) was rearranged to 4-hydroxycyclopent-2-enone (±)-137 in

    moderate yield (Scheme 31).

    Scheme 31. Synthesis of precursor for enzymatic resolution: a) KH2PO4, pH = 4.1, H2O, reflux, 2 d, 40%; b) TBDMSCl (1.15 eq.), NEt3 (1.50 eq.), DMAP (5 mol%), THF, 0 °C - rt, 89%; c) LiAlH4 (0.70 eq.), LiI (0.50 eq.), toluene/TBME, -30 °C, 3 h, 85% (cis/trans 92:8).

    The following TBDMS-protection of the free hydroxy group introduces a bulky substituent in

    (±)-138, which directs the subsequent reduction towards the cis-substituted (±)-139 in good

    yield and selectivity.

    The racemate of (±)-139 was subjected to kinetic enzymatic resolution using porcine pancreas

    lipase (PPLE).[171] (-)-139 and (+)-140 were separated afterwards by simple chromatography

    on silica gel, and both compounds can be used in the further synthesis providing the important

    feature not to loose material within this early stage (Scheme 32).

    Scheme 32. Enzymatic resolution: a) porcine pancreas lipase PPLE, vinylacetate (4.50 eq.), NEt3 (0.68 eq.), TBME, rt, 48 h, (-)-139 (95%, 92% ee), (+)-140 (80%, >99% ee).

  • Main Part

    42

    The progress in the enzymatic resolution can easily be monitored via chiral GC analysis by

    determination of the ee-value for the starting material (-)-139 (Figure 10).

    Figure 10. Kinetic resolution of (-)-139 monitored by chiral GC.

    In the first 12 h the reaction velocity was high, illustrated in the steep slope of the curve at the

    beginning (Figure 10). The maximum ee of 92% for (-)-139 was reached after 48 h. A change

    of the high ee-value of >99% for the acetylated product (+)-140 was not observed during this

    reaction. All attempts to recycle the used enzyme for further resolution reactions were not

    successful and only very low conversion was seen.

    For the further synthesis, the acetyl-protection of (+)-140 was removed by saponification

    using LiOH (Scheme 33).

    OR

    OTBDMS

    OPMB

    OTBDMS

    a

    b c

    OPMB

    OH

    (+)-140: R = Ac(+)-139: R = H

    OPMB

    OH

    =

    (+)-141 (-)-142

    Scheme 33. a) LiOH (1.20 eq.) THF:MeOH:H2O (3:1:1), rt, 2 h, 96%; b) NaH (1.25 eq.), NaI (1.00 eq.), p-methoxybenzylbromide (1.30 eq.), THF, rt, 5 h, 86%; c) TBAF (1.00 eq.), NEt3 (0.10 eq.), THF, rt, 24 h, 85%.

    0

    10

    20

    30

    40

    50

    60

    70

    80

    90

    100

    0 12 24 36 48t [h]

    % ee

    for (-)-139

  • Main Part

    43

    Because TBDMS- and benzyl-protection approaches already failed in previous attempts

    within similar syntheses of guaianolides carried out in our group,[172] the strategy within this

    work was changed to the protecting group p-methoxybenzyl (PMB) which is stable to all

    conditions needed in the further synthesis and is readily removed under mild oxidative

    conditions (preferably DDQ). Protection of (+)-139 by p-methoxybenzylbromide afforded

    PMB-protected (+)-141 in 86% yield and standard TBDMS removal by TBAF led to (-)-142.

    (-)-142 is also accessible by protecting group transformation starting from (-)-139. Reacting

    the free hydroxy functionality in (-)-139 with acetic anhydride in pyridine afforded the fully

    protected compound (-)-140 in excellent yield (Scheme 34).

    OR

    OTBDMS

    OAc

    OH

    c

    OPMB

    OAc

    (-)-139: R = H(-)-140: R = Ac a

    b d

    OPMB

    OH

    (+)-143 (-)-142(+)-144

    Scheme 34. a) pyridine (15.0 eq.), Ac2O (4.5 eq.), rt, 6 h, 97%; b) TBAF (1.0 eq), NEt3 (0.1 eq.), THF, rt, 2 h, 95%; c) p-methoxybenzyltrichloroacetimidate (1.67 eq.), Cu(OTf)2 (5 mol%), CH2Cl2, 0 °C - rt, 24 h, 83%; d) LiOH (1.2 eq.), THF/MeOH/H2O (3:1:1), rt, 2 h, 92%.

    The removal of the TBDMS protection under standard conditions smoothly provided (+)-143

    as a colorless solid, which can be easily recrystallized from diethylether.

    The introduction of the required PMB protecting group proved to be difficult on this

    substrate. First attempts using PMB-Br (similar to the synthesis of (+)-141, Scheme 33) failed

    due to racemization under these conditions. However, applying the trichloroacetimidate

    protocol under Cu(OTf)2 catalysis as described by Basu et al.[173] afforded (+)-144 in good

    yield and purity. Removal of the acetyl protection by standard saponification also provided

    (-)-142 in 92% yield.

    Altogether, both compounds resulting from the enzymatic resolution can be transformed into

    the same intermediate (-)-142, providing the important possibility to assemble large quantities

    of this material on this early stage of synthesis.

  • Main Part

    44

    Final oxidation of (-)-142 using PCC yielded the PMB-protected cyclopentenone (-)-145

    (Scheme 35).

    Scheme 35. Oxidation of (-)-142 to key intermediate (-)-145: a) PCC (1.2 eq.), 4 Å MS, CH2Cl2, rt, 24 h, 86%.

    With this synthesis the key intermediate (-)-145 was accessible in 8-9 steps starting from

    commercially available furfuryl alcohol (136). Each step in the described sequence was a spot

    to spot reaction and could easily be monitored by TLC or GC. After optimization of the

    reaction conditions, up-scaling to 18 g batches in the enzymatic resolution was possible

    without loosing the enantiomeric purity of the resulting products.

    3.2 Synthesis of the chiral allylsilanes

    The PMB-protected cyclopentenone (-)-145 was subjected to a highly selective 1,4-addition

    with appropriate cuprate reagents to introduce the functionalized carbon side chain at

    C3-position.

    The desired hydroxy functionality is introduced in this step, masked as a bulky silyl group,

    offering certain advantages (Scheme 36): The desired cuprate reagents 147 with an n-electron

    donor atom (here oxygen) attached to the metalated carbon are not readily accessible from the

    corresponding halides 146 by usual procedures[174] and often involve multiple steps in their

    preparation (e.g. using tin-containing intermediates).[174,175]

    Scheme 36. Metal organyls used for 1,4-addition: a) Mg (3.0 eq.), THF, rt, 2 h.

  • Main Part

    45

    Additionally, the introduction of a further hydroxy protecting group at this early stage of the

    synthesis was thought to be problematic in terms of restrictions in transformations and in

    protecting group differentiation.

    To avoid these problems, commercially available chloromethylsilanes 148 (R = OiPr, Ph)

    were used for standard Grignard formation. In situ conversion to the appropriate cuprates

    provided the reagents for the 1,4-addition onto cyclopentenone (-)-145. The use of different

    substituents at the silicon atom (R = OiPr, Ph) opens the possibility to investigate different

    conversion protocols for the transformation of the masked hydroxy group into the

    corresponding free alcohol.

    Because the bulky PMB-protecting group in (-)-145 shields the lower half space, the cuprate

    addition proceeds highly diastereoselectively from the upper face resulting in the desired anti-

    substitution on the cyclopentane ring. The resulting enolates were trapped in the presence of

    TMS-Cl as the corresponding silylenolethers 150 and 151 (Scheme 37). These materials are

    very sensitive to heat and traces of acid.[176] Therefore, purification by distillation or

    chromatography was not possible, however, after extensive extraction the products possessed

    sufficient purity to carry on with the next steps.

    OPMB

    O

    SiMe2R

    TMS

    OPMBSiMe2

    R

    TMS

    a b(-)-145

    150 R = Ph151 R = OiPr

    152 R = Ph153 R = OiPr

    Scheme 37. Synthesis of chiral allylsilanes: a) LiCl (0.3 eq.), CuI (0.15 eq.), TMSCl (4.0 eq.), R = Ph: PhMe2SiCH2MgCl (1N in THF) (1.25 eq.), THF, -78 °C, 3 h, 99%, dr >99:1; R = OiPr: iPrOMe2SiCH2MgCl (1 N in THF) (1.15 eq.), THF, -78 °C, 3 h, 90%, dr >99:1; b) Ni(acac)2 (0.1-1.0 eq.) Me3SiCH2MgCl (1 N in Et2O) (1.5-2.0 eq.), Et2O, rt, 5 d, R = Ph: 40%, R = OiPr: 34%.

    For the final transformation of the silylenolethers into the corresponding allylsilanes 152 and

    153 a modified procedure reported by Kumada et al. was used.[177] Ni(acac)2 catalyzes the

    coupling of silylenolethers with the appropriate Grignard reagent to afford the desired

    allylsilanes 152-153 only in moderate yields (34-40%). This might be due to the highly

    substituted cyclopentane ring and the resulting steric strain preventing the cross coupling to

    the allylsilanes. Attempts to improve this reaction by gentle warming, elongated reaction

    times or increasing the catalyst loading as well as reagent excess did not lead to better results.

  • Main Part

    46

    From the Ni(acac)2 coupling reaction compounds 154 and 155 were also isolated as

    decomposition products of the corresponding silylenolether (Scheme 38).

    Scheme 38. Possible use of by-products.

    These trans-substituted cyclopentenones 154 and 155 are very interesting structures

    themselves, since sugar-like trisubstituted cyclopentanes 157 and 158 could become

    accessible within a short sequence, including Tamao-Fleming oxidation of the side chain and

    reduction of the resulting ketone 156. These pseudo-anomeric structures have already proven

    to be important compounds as sugar mimics for pharmaceutical or biochemical purposes.[178]

  • Main Part

    47

    4. Synthesis of the cyclopropylcarbaldehyde Both enantiomers of the highly functionalized 1,2,3-trisubstituted cyclopropylcarbaldehyde

    130 are readily accessible in enantiomeric pure form via a well established two step sequence

    starting from methyl-2-furoate (131).[6,74,179,180]

    The stereochemical outcome of the sequence of cyclopropanation, ozonolysis to the

    cyclopropylcarbaldehyde 130 and subsequent allylation/retroaldol-lactonization depends on

    the stereochemistry of the bis(oxazoline)-ligand (BOX) 159 initially used (Figure 11 and

    Scheme 39).

    Figure 11. iPr-BOX-ligands (+)/(-)-159 used for asymmetric cyclopropanation.

    Scheme 39. Stereochemical relationships of iPr-BOX-ligands and stereochemistry of lactone aldehydes.

    The (R,R)-iPr-BOX ligand (+)-159 derived from D-valine leads to the desired substitution

    pattern on the lactone aldehyde 161 as found in the natural products, and subsequently the

    (S,S)-iPr-BOX ligand (-)-159, prepared from the less expensive natural L-valine, gives rise to

    the enantiomeric compound 162.

  • Main Part

    48

    Both enantiomers of the chiral BOX-ligands 159 were prepared starting from D- or L-valinol

    164, derived from the corresponding amino acids by reduction with sodiumborohydride and

    iodine. Coupling of 2,2-dimethylpropane-dioyl-dichloride 163 and valinol 164 proceeded

    successfully to diamide 165 (Scheme 40). Subsequent tosylation and cyclization gives rise to

    the corresponding enantiomeric pure ligand (+)-159.[181]

    Scheme 40. Synthesis of bis(4-isopropyloxazoline) ligand: a) valinol (2.0 eq.), NEt3 (2.5 eq.), CH2Cl2, 0-rt °C, 70 min, 84%; b) DMAP (10 mol%), NEt3 (4.0 eq.), TsCl (2.0 eq.), CH2Cl2, rt, 27 h, 83%.

    BOX-ligand (+)-159 was applied in a Cu(I)-mediated asymmetric regio- and

    diastereoselective cyclopropanation of methyl-2-furoate (131), resulting in (+)-160 with high

    enantioselectivity of 85-90% ee, which was improved to >99% ee after recrystallization

    (Scheme 41).

    Scheme 41. Cyclopropanation and ozonolysis: a) (i) ethyl diazoacetate (2.67 eq.), Cu(OTf)2 (0.66 mol%), (+)-159 (0.84 mol%), PhNHNH2 (0.70 mol%), CH2Cl2, 0 °C, 54%, 85-90% ee; (ii) recrystallization (CH2Cl2, pentane), >99% ee, 37%; b) (i) O3, CH2Cl2, -78 °C; (ii) DMS (4.00 eq.), 22 h, -78 °C - rt, 90%.

    The stereochemical outcome of this reaction can be explained applying the models suggested

    by Pfaltz[182] and Andersson[183] for the asymmetric cyclopropanation of alkenes:

    The reactive complex 166 in this reaction is shown in Figure 12 (left). An approach of 131

    (substituent CO2Me oriented away from 166 to minimize steric interactions) from the right

    side is expected to be favored, since an attack from the left side shows strong repulsive steric

    interaction of the approaching olefin 131 and the iPr group of the ligand (+)-159 (Figure 12,

    right). An attack from the right side will also lead to a flipping of the ester group E in 166 to

    the left (counterclockwise), resulting in only small interactions with the hydrogen

    substituent Ha.

  • Main Part

    49

    Figure 12. Reactive complex and model for asymmetric cyclopropanation.

    In the subsequent cyclopropanation the less substituted and presumably more electron rich

    double bond of 131 is attacked.

    The formation of the corresponding endo-diastereomer was not observed and after

    optimization of the reaction conditions a scale-up to 50-100 g was possible, still affording

    high enantioselectivity, so that both enantiomers of 160 can be prepared in optical pure form

    and multigram quantities.

    To access the cyclopropylcarbaldehyde, the double bond present in (+)-160 was cleaved by

    ozonolysis followed by reductive work-up leading to (+)-130 in 90% yield (Scheme 41). This

    material rendered solid upon treatment with diethylether and was stable for months while

    stored under a nitrogen atmosphere at -35 °C.

  • Main Part

    50

    5. Formation of the anti-substituted lactone aldehyde The next step was the addition of the chiral allylsilanes to the cyclopropylcarbaldehyde. In

    general, the stereocontrol of additions onto a cyclopropyl-substituted carbonyl compound 167

    can be explained by analyzing the conformational preferences and applying the Felkin-Anh-

    model[184] in combination with the Curtin-Hammett-principle.[185]

    The cyclopropane ring shows in analogy to an alkene double bond strong π-donating

    properties. These are only effective in the two bisected conformations, which are very similar

    to the Felkin-Anh conformations s-cis-167 and s-trans-167 of the cyclopropyl-substituted

    carbonyl compounds (Scheme 42).

    OH

    H

    OR

    CO2EtH

    H

    Nu

    (s-cis)-167 (s-trans)-167

    Nu

    OH

    CO2Et

    RO

    anti-Felkin-Anh-168

    Nu

    OH

    CO2Et

    RO

    Felkin-Anh-168

    O H

    H

    OR

    CO2EtH

    H

    R = C(O)CO2MeNu

    Scheme 42. Nucleophilic attack on cyclopropyl-substituted carbonyl compounds.

    From these possible conformations the latter is energetically more favored because the steric

    interactions of the aldehyde and the space demanding cyclopropane ring are minimized. A

    nucleophile attacking the more stable s-trans-167 conformation would consequently have to

    approach over a bulky substituent leading to the anti-Felkin-Anh-168 product. The

    experimentally observed Felkin-Anh-168 product results from the s-cis-167 conformation

    which is attacked by the nucleophile coming over a less hindered half space.

    If the activation energies of the selectivity determining step are higher than the rotation barrier

    between the different conformers, the Curtin-Hammet principle can be applied. For this case

    not the preferred conformation of the substrate (here s-trans-167) is responsible for the

    product distribution, but the activation energy of the transition state determines the

    stereochemical outcome.

    In the case described here, the rotation barrier for the transformation of s-cis to s-trans must

    be smaller than the activation barrier for the selective addition step and so the attack of bulky

    nucleophiles will occur on the less hindered path over the smaller substituent leading to the

    experimentally observed Felkin-Anh-168 product.

  • Main Part

    51

    The synthesized chiral silylenolethers 150/151 and allylsilanes 152/153 were now combined

    in a Lewis acid mediated Mukaiyama-aldol reaction or a Hosomi-Sakurai allylation with the

    cyclopropylcarbaldehyde (+)-130, respectively.

    All attempts to add the silylenolether 151 and allylsilane 153 with the OiPr- substituent at the

    silicon atom in the side chain failed due to decomposition of the silyl precursor.

    In contrast, the reactions using the Ph-substituted compounds 150 and 152 proceeded

    smoothly to the highly substituted intermediates 170 and 171 which were obtained as single

    diastereomers (Scheme 43). The Mukaiyama-aldol reaction of 150 (X = O) did not afford a

    clean product 170, as the silylenolether could not be purified and therefore had to be used as a

    crude material.

    X

    OPMB

    CHO

    HO X

    OPMBSiMe2Ph

    +

    TMS

    SiMe2Ph

    R = C(O)CO2Me

    170: X = O171: X = CH2

    H

    150: X = O152: X = CH2

    a

    CO2Et

    CO2Et

    RO

    RO

    (+)-130

    OH

    H

    OR

    CO2EtH

    HOPMB

    Me3Si

    H

    BF3

    SiMe2Ph

    169

    Scheme 43. Addition of chiral silylenolethers and allylsilanes to the cyclopropylcarbaldehyde: a) BF3⋅OEt2 (1.1 eq.), CH2Cl2, -78 °C, 16 h, 170: X = O: 95%, dr >99:1; 171: X = CH2: 95%, dr >99:1.

    These highly functionalized materials can not be purified easily by distillation due to their

    high molecular mass or chromatography because they tend to decompose on silica gel. Simple

    extraction afforded the products in sufficient purity to proceed with the next step in the

    synthesis.

    The stereochemical outcome of this reaction can be explained by the proposed transition state

    169 (Scheme 43). In this case, the nucleophile attacks the s-cis-conformation of the carbonyl

    group in anti-orientation to its bulky substituent (CH2SiMe2Ph) leading to the trans-Felkin-

    Anh-products 170 and 171 (see discussion above).

  • Main Part

    52

    In an attempt to combine 152 and (-)-130 in the mismatched case, only a complex mixture of

    at least four compounds was observed in a very low yield, which indicates the importance of

    the double stereochemical differentiation with the chiral allylsilanes.

    Upon treatment with Ba(OH)2 in methanol only the Hosomi-Sakurai allylation product 171

    (X = CH2) rearranged to the lactone aldehyde 175 in 72% yield (Scheme 44). Attempts to use

    triethylamine or LiOH as base for this transformation failed. The reaction proceeds by

    saponification of the more labile oxalic ester, upon which a ring opening of the now

    unmasked donor-acceptor substituted cyclopropane 172 is triggered, followed by

    lactonization of 173 to give 175 as a single stereoisomer.

    Scheme 44. Retroaldol-lactonization: a) Ba(OH)2⋅8H2O (0.55 eq.), MeOH, rt, 2 h, 72%, dr >99:1; b) i) O3, CH2Cl2, -78 °C, 15 min; ii) DMS (4.0 eq.), -78 °C - rt, 24 h.

    In contrast, the addition product 170 which resulted from the Mukaiyama-aldol reaction failed

    to rearrange into the ketone-aldehyde 176. Therefore, an attempt was made to subject lactone

    aldehyde 175 to ozonolysis conditions to access the ketoaldehyde 176, but only

    decomposition of the starting material was observed.

    The lactone aldehyde 175 is an important key-intermediate in further investigations towards

    the total synthesis of guaianolide sesquiterpene lactones: The trans-stereochemistry at the

    lactone ring is already set and the cyclopentane ring is highly functionalized also having the

    stereochemistry at the connection point to the lactone ring and the protected hydroxy group

    set in the right way. The aldehyde group and the double bond present in the molecule are

    versatile functionalities, which allow many further transformations to construct the framework

    of the guaianolides and related natural products via certain ring closing reactions.

  • Main Part

    53

    6. Investigations towards 5,6,5-ring systems In addition to oxidative derivatisation, nature has also the possibility to rearrange the complex

    5,7,5-carbon skeleton of the guaianolides. Probably by a Wagner-Meerwein-type

    rearrangement a 5,6,5-ring system is formed, as for example found in Lasiolaenolide (178)

    and its derivatives, isolated from various plants, such as Lasiolaena santosii (Scheme 45).[186]

    Scheme 45. Ring closing reactions to form rearranged guaianolides.

    The lactone-aldehyde 175 should be a suitable precursor to construct the core 177 of this

    family of natural products. Connecting the carbonyl-carbon with the opposite double bond

    would lead to a 5,6,5-ring system. Although, the methyl group at the C5-position is lacking

    this transformation would provide a direct and interesting route to this skeleton.

    6.1 Intramolecular carbonyl-ene reaction

    The first approach to perform this ring closure was the utilization of an intramolecular

    carbonyl-ene reaction on aldehyde 175 which should directly lead to the 5,6,5-membered ring

    system 179 (Scheme 46).

    Scheme 46. Intramolecular carbonyl-en-reaction: a) see Table 2.

  • Main Part

    54

    Although intramolecular carbonyl-ene reactions on less functionalized precursors have been

    reported,[187] treating lactone aldehyde 175 with different Lewis acids at different

    temperatures did not lead to the desired tricyclic structure 179 (Table 2). At low temperatures

    (-78 °C) no reaction was observed and at higher temperature (0 °C or rt) only decomposition

    of the starting material occurred.

    Table 2. Intramolecular carbonyl-ene-reaction conditions.

    Entry Lewis Acid Reaction conditions Yield

    1 BF3⋅OEt2 (2.0 eq.) -78 °C, 4 h no reaction

    2 BF3⋅OEt2 (2.0 eq.) rt, 4 h decomposition

    3 BF3⋅OEt2 (2.0 eq.) 0 °C, 1 h decomposition

    4 TiCl4 (2.0 eq.) 0 °C, 1 h decomposition

    5 SnCl4 (2.0 eq.) 0 °C, 1 h decomposition

    The difficulties encountered in this transformation might be due to the high functionalization

    of 175 and moreover, the PMB protecting group might not be stable under these Lewis acidic

    conditions.

    Scheme 47. PMB deprotection of 175: a) DDQ (1.14 eq.), CH2Cl2/H2O, rt, 2 h.

    To remove the PMB protecting group, 175 was treated with DDQ (Scheme 47). This resulted

    in an inseparable mixture of unknown products and did not afford the desired free

    alcohol 180.

  • Main Part

    55

    6.2 SmI2-promoted radical cyclization

    Another potential reaction for the construction of the skeleton of type 177 is the SmI2

    promoted intramolecular radical cyclization of aldehydes into C=C-double bonds which

    already has found many applications in the total synthesis of various natural products.[188]

    SmI2 is a one-electron-transfer reagent, which should transform the lactone aldehyde 175 into

    the radical intermediate 181 (Scheme 48). Subsequent cyclization by incorporating the

    exo-double bond would led to the 5,6,5-membered ring system 182. The 6-endo product

    should form with preference rather than the 5-exo product, since the trans-configuration at the

    lactone moiety introduces strain into the system. This should lead to the formation of the

    larger ring which is presumably thermodynamically more stable. Moreover, upon the 5-exo

    ring closure a primary radical rather than a tertiary radical as for the 6-endo ring closure

    would be formed.

    Scheme 48. SmI2-promoted radical cyclization: a) SmI2 (2.0 eq.), THF, 0 °C, 2 h.

    Unfortunately, applying the standard SmI2 cyclization conditions on lactone aldehyde 175 did

    not afford the expected tricyclic structure and resulted in an inseparable mixture of

    undefinable products.

  • Main Part

    56

    7. Investigations towards the guaianolide core skeleton Since all previous attempts (carbonyl-ene reaction and SmI2-radical cyclization) to build a

    tricyclic framework failed, the strategy was changed, focusing on the construction of the

    5,7,5-membered ring system of the guaianolides. Different approaches to close the central

    7-membered ring were explored including radical cyclization and ring closing metathesis.

    7.1 Radical cyclization approach

    We were already able to show the transformation of simplified precursors 71 into bi- and

    tricyclic sesquiterpene lactone skeletons 72 via radical cyclizations (Scheme 49).[74]

    Scheme 49. Synthesis of unsubstituted bi- and tricyclic sesquiterpene lactone scaffolds by radical cyclization.

    This promising sequence was investigated on lactone aldehyde 175. Alkenylation by a

    modified Horner-Wadsworth-Emmons (HWE) reaction gave rise to 183 in 72% yield and in

    an expected preference for the Z-isomer (E/Z = 17:83). Here it has to be noted that it proved

    to be more convenient and higher yielding to generate the 2-bromo-phosphonoacetate in situ

    to prevent the formation of unhalogenated HWE-product.

    Scheme 50. Radical cyclizations towards the guaianolide skeleton: a) (i) NaH (1.05 eq.), triethylphosphono-acetate (1.05 eq.), Br2 (1.11 eq.), THF, 0 °C, 1 h; (ii) NaH (1.05 eq.), 175, THF, 0 °C to rt, 1 h, 72%, E/Z = 17:83; b) AIBN (0.2 eq.), Bu3SnH (1.5 eq.), benzene, reflux, 2 h, 89%, dr = 87:13; c) DDQ (1.3 eq.), CH2Cl2, pH 7 buffer, rt, 4 h, 89%.

  • Main Part

    57

    Under diluted radical generating conditions (AIBN, Bu3SnH) an E/Z-mixture of 183 (17:83)

    reacted to the tricyclic guaianolide scaffold 184. While comparing the remarkably high yield

    (89%) of this radical cyclization with the E/Z-ratio of the precursor it is clear, that both

    isomers can be cyclized.

    Deprotection of the PMB-group on the finalized core skeleton under standard conditions

    using DDQ gave rise to 185 in 89% yield (Scheme 50), which can be subjected to further

    functionalization (e.g. oxidation, esterification, glycosylation).

    Stork et al. have shown the synthetic value of vinyl radicals.[189,190] Based on this work it was

    recognized, that a fast inversion of the radical species between E- and the corresponding

    Z-isomer takes place. An explanation for this behavior is found in the analysis of the involved

    orbitals (Scheme 51).

    Scheme 51. Orbital inversion of vinyl radicals.

    The orbital hybridization of 186 and 188 with a bent structure is best described as sp2-hybrids:

    The unpaired electron is therefore found in a sp2-orbital and an angle of approximately 60 °

    against the elongated C-C bond is included.

    The transition state of the radical inversion is best represented with the linear structure 187

    having a sp-hybridization and the unpaired electron located in a p-orbital perpendicular to the

    π-bond. The geometry of the radical center is highly influenced by the π-(C=C)-double bond

    and the σ-(C-R3)-single bond and a nearly linear assembly is assumed. Indeed, this

    assumption was proven by EPR-spectroscopy (especially for R3 = CO2H or Ph)[191] and a low

    inversion barrier for the interconversion of Z- and E-isomers is seen, resulting in a fast

    inversion of the vinyl radical.[192,193] Therefore, both isomers can take part in the cyclization

    reaction to give rise to the observed high yield of 89% in this transformation.

  • Main Part

    58

    The mechanism for the cyclization reaction of 183 to 184 appears obvious, but there are two

    pathways possible, both leading to the same final product (Scheme 52):

    After initial radical formation, a reaction can take place directly with the opposite double

    bond present in 189 to form the observed product 184 via a 7-endo-trig cyclization.

    Also, an initial 6-exo-trig cyclization of 189 towards 190 is conceivable, providing a primary

    radical which can subsequently form the cyclopropinylcarbinyl radical 191. Rearrangement of

    this highly strained structure leads also to the isolated 7-endo-product 184.

    Although, radical trapping experiments in the presence of ethylacrylate as a possible

    scavenger were tried, the exact reaction mechanism remains unknown.

    Scheme 52. Possible radical cyclization mechanisms towards 184.

    The stereochemistry of the final product 184 was then determined by NOE-experiments

    where a clear coupling between 6a-3a, 6a-9a and 6a-CH2Si-protons is seen only for the major

    isomer. This is only possible in the cis-fused case, where these protons are found on the same

    side of the molecule. This preference (87:13) can be explained by a lower allover strain within

    the guaianolide core by a cis-fused cyclopentane ring system, where the final H-addition

    occurs from the top face of the molecule.

  • Main Part

    59

    7.2 Ring closing metathesis approach

    Ring closing metathesis (RCM) is a versatile tool in organic chemistry and has already proven

    t